EP2691529B1 - AAV2-Partikel, umfassend ein AAV2-Capsidprotein und einen Vektor, umfassend eine Nukleinsäure, die eine Tripeptidylpeptidase 1 (TPP1) kodiert, zur Verwendung für die Behandlung von spätinfaniler Ceroid-Lipofuszinose (LINCL) in einem Nicht-Nagetier-Säugetier durch intraventrikuläre Injektion oder ICV-Gabe. - Google Patents

AAV2-Partikel, umfassend ein AAV2-Capsidprotein und einen Vektor, umfassend eine Nukleinsäure, die eine Tripeptidylpeptidase 1 (TPP1) kodiert, zur Verwendung für die Behandlung von spätinfaniler Ceroid-Lipofuszinose (LINCL) in einem Nicht-Nagetier-Säugetier durch intraventrikuläre Injektion oder ICV-Gabe. Download PDF

Info

Publication number
EP2691529B1
EP2691529B1 EP12763234.7A EP12763234A EP2691529B1 EP 2691529 B1 EP2691529 B1 EP 2691529B1 EP 12763234 A EP12763234 A EP 12763234A EP 2691529 B1 EP2691529 B1 EP 2691529B1
Authority
EP
European Patent Office
Prior art keywords
aav2
vector
nucleic acid
cell
aav
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
EP12763234.7A
Other languages
English (en)
French (fr)
Other versions
EP2691529A1 (de
EP2691529A4 (de
Inventor
Beverly L. Davidson
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of Iowa Research Foundation UIRF
Original Assignee
University of Iowa Research Foundation UIRF
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by University of Iowa Research Foundation UIRF filed Critical University of Iowa Research Foundation UIRF
Priority to EP19170680.3A priority Critical patent/EP3546584B1/de
Publication of EP2691529A1 publication Critical patent/EP2691529A1/de
Publication of EP2691529A4 publication Critical patent/EP2691529A4/de
Application granted granted Critical
Publication of EP2691529B1 publication Critical patent/EP2691529B1/de
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/0075Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the delivery route, e.g. oral, subcutaneous
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/48Hydrolases (3) acting on peptide bonds (3.4)
    • C12N9/485Exopeptidases (3.4.11-3.4.19)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2799/00Uses of viruses
    • C12N2799/02Uses of viruses as vector
    • C12N2799/021Uses of viruses as vector for the expression of a heterologous nucleic acid
    • C12N2799/025Uses of viruses as vector for the expression of a heterologous nucleic acid where the vector is derived from a parvovirus

Definitions

  • Gene transfer is now widely recognized as a powerful tool for analysis of biological events and disease processes at both the cellular and molecular level. More recently, the application of gene therapy for the treatment of human diseases, either inherited (e.g., ADA deficiency) or acquired ( e.g., cancer or infectious disease), has received considerable attention. With the advent of improved gene transfer techniques and the identification of an ever expanding library of defective gene-related diseases, gene therapy has rapidly evolved from a treatment theory to a practical reality.
  • gene therapy has been defined as a procedure in which an exogenous gene is introduced into the cells of a patient in order to correct an inborn genetic error.
  • human diseases are currently classified as genetic, specific mutations in the human genome have been identified for relatively few of these diseases.
  • these rare genetic diseases represented the exclusive targets of gene therapy efforts. Accordingly, most of the NIH approved gene therapy protocols to date have been directed toward the introduction of a functional copy of a defective gene into the somatic cells of an individual having a known inborn genetic error.
  • gene therapy has more recently been broadly defined as the correction of a disease phenotype through the introduction of new genetic information into the affected organism.
  • a transferred gene is introduced into cells of the recipient organism in situ that is, within the recipient.
  • In vivo gene therapy has been examined in several animal models.
  • Several recent publications have reported the feasibility of direct gene transfer in situ into organs and tissues such as muscle, hematopoietic stem cells, the arterial wall, the nervous system, and lung.
  • Direct injection of DNA into skeletal muscle, heart muscle and injection of DNA-lipid complexes into the vasculature also has been reported to yield a detectable expression level of the inserted gene product(s) in vivo.
  • LSD lysosomal storage diseases
  • CNS central nervous system
  • Watson et al. Gene Therapy, vol. 13, 2006, pages 917-925 describes the use of recombinant AAV vectors for delivering a therapeutic protein to the brain of a rodent mammal in need thereof. More specifically, Watson et al. describes the use of an rAAV-IDUA vector packaged in an AAV2 capsid for treating a lysosomal storage disease (LSD) in adult mice, wherein the LSD is mucopolysaccharidosis type I and the vector is administered by intrathecal injection.
  • LSD lysosomal storage disease
  • Passini et al. Journal of Virology, vol. 77, 2003, pages 7034-7040 describes the use of recombinant AAV vectors for delivering a therapeutic protein to the brain of a rodent mammal in need thereof, wherein the rAAV particle is administered by intraventricular injection. More specifically, Passini et al. describes the use of an rAAV1 vector for treating a LSD in mice, wherein the therapeutic protein is beta-glucuronidase and the LSD is mucopolysaccharidosis type VII.
  • AAV2CUhCLN2 a vector comprising the human CLN2 cDNA encoding the human TPP1 gene used for gene therapy of classical late infantile neuronal ceroid lipofuscinosis (cLINCL) in CLN2-/- mice.
  • the AAV2CUhCLN2 vector was stereotaxically injected and resulted in 'a marked reduction of autofluorescent storage in the AAV2CUhCLN2 injected brain regions, as well as adjacent regions, including the striatum and hippocampus'.
  • the present invention relates to an rAAV2 particle comprising an AAV2 capsid protein and a vector comprising a nucleic acid encoding a therapeutic protein inserted between a pair of AAV inverted terminal repeats for use in treating a lysosomal storage disease (LSD) in a non-rodent mammal, wherein the rAAV2 particle is to be administered to the non-rodent mammal by intraventricular injection or ICV delivery, wherein the protein is tripeptidyl peptidase 1 (TPP1), wherein the LSD is late infantile ceroid lipofuscinoses (LINCL), and wherein the non-rodent mammal is a dog, primate or human.
  • LSD lysosomal storage disease
  • TPP1 tripeptidyl peptidase 1
  • LINCL late infantile ceroid lipofuscinoses
  • Disclosed is a method of delivering a nucleic acid to an ependymal cell of a non-rodent mammal comprising administering to the ependymal cell an AAV2 particle containing a vector comprising the nucleic acid inserted between a pair of AAV2 inverted terminal repeats, thereby delivering the nucleic acid to the ependymal cell.
  • the rAAV2 particle infects the non-primate ependymal cell at an rate of more than 20% than the infectivity rate of AAV4, such as at a rate of more than 50% or 100%, 1000% or 2000% than the infectivity rate of AAV4.
  • a method of delivering a nucleic acid to a non-rodent mammal comprising administering to an ependymal cell from the mammal an AAV2 particle comprising the nucleic acid inserted between a pair of AAV inverted terminal repeats, and returning the ependymal cell to the mammal, thereby delivering the nucleic acid to the mammal.
  • a method of delivering a nucleic acid to an ependymal cell in a non-rodent mammal comprising administering to the mammal an AAV2 particle comprising the nucleic acid inserted between a pair of AAV inverted terminal repeats, thereby delivering the nucleic acid to an ependymal cell in the mammal.
  • a method to deliver an agent to the central nervous system of a non-rodent mammal comprising administering to the cerebrospinal fluid (CSF) of the non-rodent mammal an AAV2 particle containing a vector comprising the nucleic acid inserted between a pair of AAV inverted terminal repeats in a manner effective to infect ependymal cells in the non-rodent mammal such that the ependymal cells secret the agent into the CSF of the non-rodent mammal.
  • CSF cerebrospinal fluid
  • a method of treating a disease in a non-rodent mammal comprising administering to the ependymal cells of the mammal an AAV2 particle containing a vector comprising the nucleic acid inserted between a pair of AAV inverted terminal repeats, thereby delivering the nucleic acid to the ependymal cell.
  • the disease is a lysosomal storage disease (LSD).
  • LSD is infantile or late infantile ceroid lipofuscinoses, neuronopathic Gaucher, Juvenile Batten, Fabry, MLD, Sanfilippo A,, Hunter, Krabbe, Morquio, Pompe, Niemann-Pick C, Tay-Sachs, Hurler (MPS-I H), Sanfilippo B, Maroteaux-Lamy, Niemann-Pick A, Cystinosis, Hurler-Scheie (MPS-I H/S), Sly Syndrome (MPS VII), Scheie (MPS-I S), Infantile Batten, GM1 Gangliosidosis, Mucolipidosis type II/III, or Sandhoff disease.
  • the disease is LINCL.
  • the disease is a neurodegenerative disease, such as Huntington's disease, ALS, hereditary spastic hemiplegia, primary lateral sclerosis, spinal muscular atrophy, Kennedy's disease, Alzheimer's disease, a polyglutamine repeat disease, or Parkinson's disease.
  • the large mammal is a primate, horse, sheep, goat, pig, or dog.
  • the primate is a human.
  • nucleic acid is a lysosomal hydrolase. In certain embodiments, the nucleic acid is TPP1.
  • a method of transfecting an ependymal cell a non-rodent mammalian brain comprising administering to the cerebrospinal fluid (CSF) of the non-rodent mammal an AAV2 particle containing a vector comprising a nucleic acid inserted between a pair of AAV2 inverted terminal repeats in a manner effective to infect ependymal cells in the non-rodent mammal such that the ependymal cells secrete the agent into the CSF of the non-rodent mammal.
  • CSF cerebrospinal fluid
  • the present invention relates to an rAAV2 particle comprising an AAV2 capsid protein and a vector comprising a nucleic acid encoding a therapeutic protein inserted between a pair of AAV inverted terminal repeats for use in treating a lysosomal storage disease (LSD) in a non-rodent mammal, wherein the rAAV2 particle is to be administered to the non-rodent mammal by intraventricular injection or ICV delivery, wherein the protein is tripeptidyl peptidase 1 (TPP1), wherein the LSD is late infantile ceroid lipofuscinoses (LINCL), and wherein the non-rodent mammal is a dog, primate or human.
  • LSD lysosomal storage disease
  • TPP1 tripeptidyl peptidase 1
  • LINCL late infantile ceroid lipofuscinoses
  • Adeno associated virus is a small nonpathogenic virus of the parvoviridae family. AAV is distinct from the other members of this family by its dependence upon a helper virus for replication. In the absence of a helper virus, AAV may integrate in a locus specific manner into the q arm of chromosome 19.
  • the approximately 5 kb genome of AAV consists of one segment of single stranded DNA of either plus or minus polarity. The ends of the genome are short inverted terminal repeats which can fold into hairpin structures and serve as the origin of viral DNA replication. Physically, the parvovirus virion is non-enveloped and its icosohedral capsid is approximately 20 nm in diameter.
  • AAV2 Adeno-Associated Virus Serotype 4
  • J. Vir., 80 (23):11556-11570 2006
  • the genome of AAV2 is 4680 nucleotides in length and contains two open reading frames (ORFs).
  • the left ORF encodes the non-structural Rep proteins, Rep 40, Rep 52, Rep 68 and Rep 78, which are involved in regulation of replication and transcription in addition to the production of single-stranded progeny genomes.
  • Rep proteins have been associated with the preferential integration of AAV genomes into a region of the q arm of human chromosome 19.
  • Rep68/78 has also been shown to possess NTP binding activity as well as DNA and RNA helicase activities.
  • the Rep proteins possess a nuclear localization signal as well as several potential phosphorylation sites. Mutation of one of these kinase sites resulted in a loss of replication activity.
  • ITR inverted terminal repeats
  • GAGC GAGC repeat motif
  • trs terminal resolution site
  • the repeat motif has been shown to bind Rep when the ITR is in either a linear or hairpin conformation. This binding serves to position Rep68/78 for cleavage at the trs which occurs in a site- and strand-specific manner.
  • Rep binding site is a Rep binding site with an adjacent trs.
  • the AAV2 virion is a non-enveloped, icosohedral particle approximately 25 nm in diameter, consisting of three related proteins referred to as VP1, VP2 and VP3.
  • the right ORF encodes the capsid proteins VP1, VP2, and VP3. These proteins are found in a ratio of 1:1:10 respectively and are all derived from the right-hand ORF.
  • the capsid proteins differ from each other by the use of alternative splicing and an unusual start codon. Deletion analysis has shown that removal or alteration of VP1 which is translated from an alternatively spliced message results in a reduced yield of infections particles. Mutations within the VP3 coding region result in the failure to produce any single-stranded progeny DNA or infectious particles.
  • An AAV2 particle is a viral particle comprising an AAV2 capsid protein.
  • An AAV2 capsid polypeptide can encode the entire VP1, VP2 and VP3 polypeptide.
  • the particle can be a particle comprising AAV2 and other AAV capsid proteins (i.e., a chimeric protein, such as AAV4 and AAV2). Variations in the amino acid sequence of the AAV2 capsid protein are contemplated herein, as long as the resulting viral particle comprises the AAV2 capsid remains antigenically or immunologically distinct from AAV4, as can be routinely determined by standard methods. Specifically, for example, ELISA and Western blots can be used to determine whether a viral particle is antigenically or immunologically distinct from AAV4. Furthermore, the AAV2 viral particle preferably retains tissue tropism distinct from AAV4.
  • An AAV2 particle is a viral particle comprising an AAV2 capsid protein.
  • An AAV2 capsid polypeptide encoding the entire VP1, VP2, and VP3 polypeptide can overall have at least about 63% homology (or identity) to the polypeptide having the amino acid sequence encoded by nucleotides set forth in SEQ ID NO:1 (AAV2 capsid protein).
  • the capsid protein can have about 70% homology, about 75% homology, 80% homology, 85% homology, 90% homology, 95% homology, 98% homology, 99% homology, or even 100% homology to the protein set forth in SEQ ID NO:1.
  • the capsid protein can have about 70% identity, about 75% identity, 80% identity, 85% identity, 90% identity, 95% identity, 98% identity, 99% identity, or even 100% identity to the protein set forth in SEQ ID NO:1.
  • the particle can be a particle comprising both AAV4 and AAV2 capsid protein, i.e., a chimeric protein. Variations in the amino acid sequence of the AAV2 capsid protein are contemplated herein, as long as the resulting viral particle comprising the AAV2 capsid remains antigenically or immunologically distinct from AAV4, as can be routinely determined by standard methods. Specifically, for example, ELISA and Western blots can be used to determine whether a viral particle is antigenically or immunologically distinct from AAV4.
  • the AAV2 viral particle preferably retains tissue tropism distinction from AAV4, such as that exemplified in the examples herein, though an AAV2 chimeric particle comprising at least one AAV2 coat protein may have a different tissue tropism from that of an AAV2 particle consisting only of AAV2 coat proteins.
  • AAV2 capsid sequence and AAV4 capsid sequence are about 60% homologous.
  • the AAV2 capsid comprises (or consists of) a sequence that is at least 65% homologous to the amino acid sequence set forth in SEQ ID NO:1.
  • an AAV2 particle containing, i.e., encapsidating, a vector comprising a pair of AAV2 inverted terminal repeats.
  • the nucleotide sequence of AAV2 ITRs is known in the art.
  • the particle can be a particle comprising both AAV4 and AAV2 capsid protein, i.e., a chimeric protein.
  • the particle can be a particle encapsidating a vector comprising a pair of AAV inverted terminal repeats from other AAVs (e.g., AAV1-AAV8).
  • the vector encapsidated in the particle can further comprise an exogenous nucleic acid inserted between the inverted terminal repeats.
  • AAV vectors have been shown in vitro to stably integrate into the cellular genome; possess a broad host range; transduce both dividing and non dividing cells in vitro and in vivo and maintain high levels of expression of the transduced genes.
  • Viral particles are heat stable, resistant to solvents, detergents, changes in pH, temperature, and can be concentrated on CsCl gradients. Integration of AAV provirus is not associated with any long term negative effects on cell growth or differentiation.
  • the ITRs have been shown to be the only cis elements required for replication, packaging and integration and may contain some promoter activities.
  • An AAV2 particle is a viral particle comprising an AAV2 capsid protein.
  • a recombinant AAV2 vector is a nucleic acid construct that comprises at least one unique nucleic acid of AAV2.
  • a recombinant AAV2 virion is a particle containing a recombinant AAV2 vector.
  • AAV2 ITRs the nucleotide sequence must retain one or both features described herein that distinguish the AAV2 ITR from the AAV4 ITR: (1) three (rather than four as in AAV4) "GAGC” repeats and (2) in the AAV2 ITR Rep binding site the fourth nucleotide in the first two "GAGC” repeats is a C rather than a T.
  • the promoter can be any desired promoter, selected by known considerations, such as the level of expression of a nucleic acid functionally linked to the promoter and the cell type in which the vector is to be used. Promoters can be an exogenous or an endogenous promoter. Promoters can include, for example, known strong promoters such as SV40 or the inducible metallothionein promoter, or an AAV promoter, such as an AAV p5 promoter.
  • promoters include promoters derived from actin genes, immunoglobulin genes, cytomegalovirus (CMV), adenovirus, bovine papilloma virus, adenoviral promoters, such as the adenoviral major late promoter, an inducible heat shock promoter, respiratory syncytial virus, Rous sarcomas virus (RSV), etc.
  • the promoter can be AAV2 p5 promoter or AAV4 p5 promoter.
  • fragments of p5 promoter that retain promoter activity can readily be determined by standard procedures including, for example, constructing a series of deletions in the p5 promoter, linking the deletion to a reporter gene, and determining whether the reporter gene is expressed, i.e., transcribed and/or translated.
  • the AAV2 vector can further comprise an exogenous (heterologous) nucleic acid functionally linked to the promoter.
  • heterologous nucleic acid is meant that any heterologous or exogenous nucleic acid can be inserted into the vector for transfer into a cell, tissue or organism.
  • the nucleic acid can encode a polypeptide or protein or an antisense RNA, for example.
  • functionally linked is meant such that the promoter can promote expression of the heterologous nucleic acid, as is known in the art, such as appropriate orientation of the promoter relative to the heterologous nucleic acid.
  • the heterologous nucleic acid preferably has all appropriate sequences for expression of the nucleic acid, as known in the art, to functionally encode, i.e., allow the nucleic acid to be expressed.
  • the nucleic acid can include, for example, expression control sequences, such as an enhancer, and necessary information processing sites, such as ribosome binding sites, RNA splice sites, polyadenylation sites, and transcriptional terminator sequences.
  • the heterologous nucleic acid can encode beneficial proteins that replace missing or defective proteins required by the subject into which the vector in transferred or can encode a cytotoxic polypeptide that can be directed, e.g., to cancer cells or other cells whose death would be beneficial to the subject.
  • the heterologous nucleic acid can also encode antisense RNAs that can bind to, and thereby inactivate, mRNAs made by the subject that encode harmful proteins.
  • antisense polynucleotides can be produced from a heterologous expression cassette in an AAV2 viral construct where the expression cassette contains a sequence that promotes cell-type specific expression.
  • heterologous nucleic acids which can be administered to a cell or subject as part of the present AAV2 vector can include, but are not limited to the nucleic acids encoding therapeutic agents, such as lysosomal hydrolases; tumor necrosis factors (TNF), such as TNF-alpha; interferons, such as interferon-alpha, interferon-beta, and interferon-gamma; interleukins, such as IL-1, IL-1beta, and ILs-2 through -14; GM-CSF; adenosine deaminase; secreted factors such as growth factors; ion channels; chemotherapeutics; lysosomal proteins; anti-apoptotic gene products; proteins promoting neural survival such as glutamate receptors and growth factors; cellular growth factors, such as lymphokines; soluble CD4; Factor VIII; Factor IX; T-cell receptors; LDL receptor; ApoE; ApoC; alpha-1 antitry
  • An AAV2 particle is a viral particle comprising an AAV2 capsid protein. Variations in the amino acid sequence of the AAV2 capsid protein are contemplated herein, as long as the resulting viral particle comprising the AAV2 capsid remains antigenically or immunologically distinct from AAV4, as can be routinely determined by standard methods. Specifically, for example, ELISA and Western blots can be used to determine whether a viral particle is antigenically or immunologically distinct from other AAV serotypes.
  • polypeptide refers to a polymer of amino acids and includes full-length proteins and fragments thereof.
  • protein polypeptide
  • peptide are often used interchangeably herein. Substitutions can be selected by known parameters to be neutral.
  • polypeptides having slight variations in amino acid sequences or other properties Such variations may arise naturally as allelic variations (e.g. due to genetic polymorphism) or may be produced by human intervention (e.g., by mutagenesis of cloned DNA sequences), such as induced point, deletion, insertion and substitution mutants.
  • Minor changes in amino acid sequence are generally preferred, such as conservative amino acid replacements, small internal deletions or insertions, and additions or deletions at the ends of the molecules. These modifications can result in changes in the amino acid sequence, provide silent mutations, modify a restriction site, or provide other specific mutations.
  • a method of delivering a nucleic acid to a cell comprising administering to the cell an AAV2 particle containing a vector comprising the nucleic acid inserted between a pair of AAV inverted terminal repeats, thereby delivering the nucleic acid to the cell.
  • Administration to the cell can be accomplished by any means, including simply contacting the particle, optionally contained in a desired liquid such as tissue culture medium, or a buffered saline solution, with the cells.
  • the particle can be allowed to remain in contact with the cells for any desired length of time, and typically the particle is administered and allowed to remain indefinitely.
  • the virus can be administered to the cell by standard viral transduction methods, as known in the art and as exemplified herein.
  • Titers of virus to administer can vary, particularly depending upon the cell type, but will be typical of that used for AAV transduction in general. Additionally the titers used to transduce the particular cells in the present examples can be utilized.
  • the cells can include any desired cell in humans as well as other large (non-rodent) mammals, such as primates, horse, sheep, goat, pig, and dog.
  • the present disclosure provides a method of delivering a nucleic acid to an ependymal cell, comprising administering to the ependymal cell an AAV2 particle containing a vector comprising the nucleic acid inserted between a pair of AAV inverted terminal repeats, thereby delivering the nucleic acid to the ependymal cell.
  • AAV ITRs can be AAV2 ITRs.
  • cells are isolated from a subject by standard means according to the cell type and placed in appropriate culture medium, again according to cell type. Viral particles are then contacted with the cells as described above, and the virus is allowed to transfect the cells. Cells can then be transplanted back into the subject's body, again by means standard for the cell type and tissue. If desired, prior to transplantation, the cells can be studied for degree of transfection by the virus, by known detection means and as described herein.
  • a method of delivering a nucleic acid to a cell in a subject comprising administering to the subject an AAV2 particle comprising the nucleic acid inserted between a pair of AAV inverted terminal repeats, thereby delivering the nucleic acid to a cell in the subject.
  • Administration can be an ex vivo administration directly to a cell removed from a subject, such as any of the cells listed above, followed by replacement of the cell back into the subject, or administration can be in vivo administration to a cell in the subject.
  • cells are isolated from a subject by standard means according to the cell type and placed in appropriate culture medium, again according to cell type. Viral particles are then contacted with the cells as described above, and the virus is allowed to transfect the cells.
  • Cells can then be transplanted back into the subject's body, again by means standard for the cell type and tissue. If desired, prior to transplantation, the cells can be studied for degree of transfection by the virus, by known detection means and as described herein.
  • Also disclosed is a method of delivering a nucleic acid to an ependymal cell in a subject comprising administering to the subject an AAV2 particle comprising the nucleic acid inserted between a pair of AAV inverted terminal repeats, thereby delivering the nucleic acid to an ependymal cell in the subject.
  • the amino acid sequence that targets brain vascular endothelium targets brain vascular endothelium in a subject that has a disease, e.g., a lysosomal storage disease.
  • the amino acid sequence that targets brain vascular endothelium targets brain vascular endothelium in a subject that does not have a lysosomal storage disease.
  • the viral vector comprises a nucleic acid sequence encoding a therapeutic agent.
  • the therapeutic agent is TPP1.
  • Certain aspects of the present disclosure provide a cell comprising a viral vector as described herein.
  • the cell is a mammalian cell of a non-rodent mammal. In certain embodiments, the cell is a primate cell. In certain embodiments, the cell is a human cell. In certain aspects, the cell is a non-human cell. In certain aspects, the cell is in vitro. In certain embodiments, the cell is in vivo. In certain aspects, the cell is an ependymal cell.
  • Certain aspects of the present disclosure provide a method of treating a disease in a mammal comprising administering a viral vector or the cell as described herein to the mammal.
  • the mammal is human.
  • the disease is a lysosomal storage disease (LSD).
  • LSD is infantile or late infantile ceroid lipofuscinoses, Gaucher, Juvenile Batten, Fabry, MLD, Sanfilippo A, Late Infantile Batten, Hunter, Krabbe, Morquio, Pompe, Niemann-Pick C, Tay-Sachs, Hurler (MPS-I H), Sanfilippo B, Maroteaux-Lamy, Niemann-Pick A, Cystinosis, Hurler-Scheie (MPS-I H/S), Sly Syndrome (MPS VII), Scheie (MPS-I S), Infantile Batten, GM1 Gangliosidosis, Mucolipidosis type II/III, or Sandhoff disease.
  • the disease is a neurodegenerative disease.
  • the neurodegenerative disease is Huntington's disease, ALS, hereditary spastic hemiplegia, primary lateral sclerosis, spinal muscular atrophy, Kennedy's disease, Alzheimer's disease, a polyglutamine repeat disease, or Parkinson's disease.
  • Certain aspects of the present disclosure provide a method to deliver an agent to the central nervous system of a subject, comprising administering to the CSF with a viral vector described herein so that the transduced ependymal cells express the therapeutic agent and deliver the agent to the central nervous system of the subject.
  • the viral vector transduces ependymal cells.
  • Certain aspects of the present disclosure provide a viral vector or cell as described herein for use in medical treatments.
  • Certain aspects of the present disclosure provide a use of a viral vector or cell as described herein to prepare a medicament useful for treating a disease, e.g., a lysosomal storage disease, in a mammal.
  • a disease e.g., a lysosomal storage disease
  • the vector may further comprise a lysosomal enzyme (e.g., a lysosomal hydrolase), a secreted protein, a nuclear protein, or a cytoplasmic protein.
  • a lysosomal enzyme e.g., a lysosomal hydrolase
  • secreted protein includes any secreted protein, whether naturally secreted or modified to contain a signal sequence so that it can be secreted.
  • Nucleic acid is "operably linked” when it is placed into a functional relationship with another nucleic acid sequence.
  • “operably linked” means that the DNA sequences being linked are contiguous. However, enhancers do not have to be contiguous. Linking is accomplished by ligation at convenient restriction sites. If such sites do not exist, the synthetic oligonucleotide adaptors or linkers are used in accordance with conventional practice. Additionally, multiple copies of the nucleic acid encoding enzymes may be linked together in the expression vector. Such multiple nucleic acids may be separated by linkers.
  • the present disclosure also provides a mammalian cell containing a vector described herein.
  • the cell may be human, and may be from brain.
  • the cell type may be a stem or progenitor cell population.
  • the present disclosure provides a method of treating a disease such as a genetic disease or cancer in a mammal by administering a polynucleotide, polypeptide, expression vector, or cell described herein.
  • the genetic disease or cancer may be a lysosomal storage disease (LSD) such as infantile or late infantile ceroid lipofuscinoses, Gaucher, Juvenile Batten, Fabry, MLD, Sanfilippo A, Late Infantile Batten, Hunter, Krabbe, Morquio, Pompe, Niemann-Pick C, Tay-Sachs, Hurler (MPS-I H), Sanfilippo B, Maroteaux-Lamy, Niemann-Pick A, Cystinosis, Hurler-Scheie (MPS-I H/S), Sly Syndrome (MPS VII), Scheie (MPS-I S), Infantile Batten, GM1 Gangliosidosis, Mucolipidosis type II/III, or Sandhoff disease.
  • LSD lysosomal storage disease
  • the genetic disease may be a neurodegenerative disease, such as Huntington's disease, ALS, hereditary spastic hemiplegia, primary lateral sclerosis, spinal muscular atrophy, Kennedy's disease, Alzheimer's disease, a polyglutamine repeat disease, or focal exposure such as Parkinson's disease.
  • a neurodegenerative disease such as Huntington's disease, ALS, hereditary spastic hemiplegia, primary lateral sclerosis, spinal muscular atrophy, Kennedy's disease, Alzheimer's disease, a polyglutamine repeat disease, or focal exposure such as Parkinson's disease.
  • Certain aspects of the disclosure relate to polynucleotides, polypeptides, vectors, and genetically engineered cells (modified in vivo ), and the use of them.
  • the disclosure relates to a method for gene or protein therapy that is capable of both systemic delivery of a therapeutically effective dose of the therapeutic agent.
  • a cell expression system for expressing a therapeutic agent in a mammalian recipient comprises a cell and an expression vector for expressing the therapeutic agent.
  • Expression vectors include, but are not limited to, viruses, plasmids, and other vehicles for delivering heterologous genetic material to cells.
  • the term "expression vector” as used herein refers to a vehicle for delivering heterologous genetic material to a cell.
  • the expression vector is a recombinant adenoviral, adeno-associated virus, or lentivirus or retrovirus vector.
  • the expression vector further includes a promoter for controlling transcription of the heterologous gene.
  • the promoter may be an inducible promoter (described below).
  • the expression system is suitable for administration to the mammalian recipient.
  • the expression system may comprise a plurality of non-immortalized genetically modified cells, each cell containing at least one recombinant gene encoding at least one therapeutic agent.
  • the cell expression system can be formed in vivo.
  • a method for treating a mammalian recipient in vivo includes introducing an expression vector for expressing a heterologous gene product into a cell of the patient in situ, such as via intravenous administration.
  • an expression vector for expressing the therapeutic agent is introduced in vivo into the mammalian recipient i.v., where the vector migrates via the vasculature to the brain.
  • a method for treating a mammalian recipient in vivo includes introducing the target protein into the patient in vivo.
  • the expression vector for expressing the heterologous gene may include an inducible promoter for controlling transcription of the heterologous gene product. Accordingly, delivery of the therapeutic agent in situ is controlled by exposing the cell in situ to conditions, which induce transcription of the heterologous gene.
  • the mammalian recipient may have a condition that is amenable to gene replacement therapy.
  • gene replacement therapy refers to administration to the recipient of exogenous genetic material encoding a therapeutic agent and subsequent expression of the administered genetic material in situ.
  • condition amenable to gene replacement therapy embraces conditions such as genetic diseases (i.e., a disease condition that is attributable to one or more gene defects), acquired pathologies (i.e., a pathological condition which is not attributable to an inborn defect), cancers and prophylactic processes (i.e., prevention of a disease or of an undesired medical condition).
  • therapeutic agent refers to any agent or material, which has a beneficial effect on the mammalian recipient.
  • therapeutic agent embraces both therapeutic and prophylactic molecules having nucleic acid or protein components.
  • the mammalian recipient has a genetic disease and the exogenous genetic material comprises a heterologous gene encoding a therapeutic agent for treating the disease.
  • the mammalian recipient has an acquired pathology and the exogenous genetic material comprises a heterologous gene encoding a therapeutic agent for treating the pathology.
  • the patient has a cancer and the exogenous genetic material comprises a heterologous gene encoding an anti-neoplastic agent.
  • the patient has an undesired medical condition and the exogenous genetic material comprises a heterologous gene encoding a therapeutic agent for treating the condition.
  • the term "lysosomal enzyme,” a “secreted protein,” a “nuclear protein,” or a “cytoplasmic protein” include variants or biologically active or inactive fragments of these polypeptides.
  • a "variant" of one of the polypeptides is a polypeptide that is not completely identical to a native protein. Such variant protein can be obtained by altering the amino acid sequence by insertion, deletion or substitution of one or more amino acid. The amino acid sequence of the protein is modified, for example by substitution, to create a polypeptide having substantially the same or improved qualities as compared to the native polypeptide. The substitution may be a conserved substitution.
  • a “conserved substitution” is a substitution of an amino acid with another amino acid having a similar side chain.
  • a conserved substitution would be a substitution with an amino acid that makes the smallest change possible in the charge of the amino acid or size of the side chain of the amino acid (alternatively, in the size, charge or kind of chemical group within the side chain) such that the overall peptide retains its spacial conformation but has altered biological activity.
  • common conserved changes might be Asp to Glu, Asn or Gln; His to Lys, Arg or Phe; Asn to Gln, Asp or Glu and Ser to Cys, Thr or Gly.
  • Alanine is commonly used to substitute for other amino acids.
  • the 20 essential amino acids can be grouped as follows: alanine, valine, leucine, isoleucine, proline, phenylalanine, tryptophan and methionine having nonpolar side chains; glycine, serine, threonine, cystine, tyrosine, asparagine and glutamine having uncharged polar side chains; aspartate and glutamate having acidic side chains; and lysine, arginine, and histidine having basic side chains.
  • amino acid changes are achieved by changing the codons of the corresponding nucleic acid sequence. It is known that such polypeptides can be obtained based on substituting certain amino acids for other amino acids in the polypeptide structure in order to modify or improve biological activity. For example, through substitution of alternative amino acids, small conformational changes may be conferred upon a polypeptide that results in increased activity. Alternatively, amino acid substitutions in certain polypeptides may be used to provide residues, which may then be linked to other molecules to provide peptide-molecule conjugates which, retain sufficient properties of the starting polypeptide to be useful for other purposes.
  • hydropathic index of amino acids in conferring interactive biological function on a polypeptide, wherein it is found that certain amino acids may be substituted for other amino acids having similar hydropathic indices and still retain a similar biological activity.
  • substitution of like amino acids may be made on the basis of hydrophilicity, particularly where the biological function desired in the polypeptide to be generated in intended for use in immunological embodiments.
  • hydrophilicity index or hydropathic index which assigns values to each amino acid
  • the variant protein has at least 50%, at least about 80%, or even at least about 90% but less than 100%, contiguous amino acid sequence homology or identity to the amino acid sequence of a corresponding native protein.
  • the amino acid sequence of the variant polypeptide corresponds essentially to the native polypeptide's amino acid sequence.
  • “correspond essentially to” refers to a polypeptide sequence that will elicit a biological response substantially the same as the response generated by the native protein. Such a response may be at least 60% of the level generated by the native protein, and may even be at least 80% of the level generated by native protein.
  • a variant may include amino acid residues not present in the corresponding native protein or deletions relative to the corresponding native protein.
  • a variant may also be a truncated "fragment" as compared to the corresponding native protein, i.e., only a portion of a full-length protein.
  • Protein variants also include peptides having at least one D-amino acid.
  • the variant protein may be expressed from an isolated DNA sequence encoding the variant protein.
  • "Recombinant” is defined as a peptide or nucleic acid produced by the processes of genetic engineering. It should be noted that it is well-known in the art that, due to the redundancy in the genetic code, individual nucleotides can be readily exchanged in a codon, and still result in an identical amino acid sequence.
  • the terms "protein,” “peptide” and “polypeptide” are used interchangeably herein.
  • the present disclosure provides methods of treating a disease in a mammal by administering an expression vector to a cell or patient.
  • an expression vector to a cell or patient.
  • a person having ordinary skill in the art of molecular biology and gene therapy would be able to determine, without undue experimentation, the appropriate dosages and routes of administration of the expression vector used in the novel methods of the present disclosure.
  • the cells are transformed or otherwise genetically modified in vivo.
  • the cells from the mammalian recipient are transformed ( i . e., transduced or transfected) in vivo with a vector containing exogenous genetic material for expressing a heterologous (e.g., recombinant) gene encoding a therapeutic agent and the therapeutic agent is delivered in situ.
  • a heterologous (e.g., recombinant) gene encoding a therapeutic agent and the therapeutic agent is delivered in situ.
  • exogenous genetic material refers to a nucleic acid or an oligonucleotide, either natural or synthetic, that is not naturally found in the cells; or if it is naturally found in the cells, it is not transcribed or expressed at biologically significant levels by the cells.
  • exogenous genetic material includes, for example, a non-naturally occurring nucleic acid that can be transcribed into anti-sense RNA, as well as a “heterologous gene” (i.e., a gene encoding a protein which is not expressed or is expressed at biologically insignificant levels in a naturally-occurring cell of the same type).
  • the mammalian recipient has a condition that is amenable to gene replacement therapy.
  • gene replacement therapy refers to administration to the recipient of exogenous genetic material encoding a therapeutic agent and subsequent expression of the administered genetic material in situ.
  • condition amenable to gene replacement therapy embraces conditions such as genetic diseases (i.e., a disease condition that is attributable to one or more gene defects), acquired pathologies ( i.e., a pathological condition which is not attributable to an inborn defect), cancers and prophylactic processes (i.e., prevention of a disease or of an undesired medical condition).
  • therapeutic agent refers to any agent or material, which has a beneficial effect on the mammalian recipient.
  • therapeutic agent embraces both therapeutic and prophylactic molecules having nucleic acid (e.g ., antisense RNA) and/or protein components.
  • the condition amenable to gene replacement therapy is a prophylactic process, i.e., a process for preventing disease or an undesired medical condition.
  • the instant disclosure embraces a cell expression system for delivering a therapeutic agent that has a prophylactic function (i.e., a prophylactic agent) to the mammalian recipient.
  • the term "therapeutic agent” includes, but is not limited to, agents associated with the conditions listed above, as well as their functional equivalents.
  • the term “functional equivalent” refers to a molecule (e.g., a peptide or protein) that has the same or an improved beneficial effect on the mammalian recipient as the therapeutic agent of which is it deemed a functional equivalent.
  • a viral vector of the disclosure is an AAV2 vector.
  • An "AAV2" vector refers to an adeno-associated virus, and may be used to refer to the naturally occurring wild-type virus itself or derivatives thereof. The term covers all subtypes, serotypes and pseudotypes, and both naturally occurring and recombinant forms, except where required otherwise.
  • serotype refers to an AAV which is identified by and distinguished from other AAVs based on capsid protein reactivity with defined antisera, e.g., there are eight known serotypes of primate AAVs, AAV-1 to AAV-8.
  • serotype AAV2 is used to refer to an AAV which contains capsid proteins encoded from the cap gene of AAV2 and a genome containing 5' and 3' ITR sequences from the same AAV2 serotype.
  • rAAV1 may be used to refer an AAV having both capsid proteins and 5'-3' ITRs from the same serotype or it may refer to an AAV having capsid proteins from one serotype and 5'-3' ITRs from a different AAV serotype, e.g., capsid from AAV serotype 2 and ITRs from AAV serotype 5.
  • rAAV refers to recombinant adeno-associated virus, also referred to as a recombinant AAV vector (or "rAAV vector").
  • AAV virus or “AAV viral particle” refers to a viral particle composed of at least one AAV capsid protein (preferably by all of the capsid proteins of a wild-type AAV) and an encapsidated polynucleotide. If the particle comprises heterologous polynucleotide (i.e., a polynucleotide other than a wild-type AAV genome such as a transgene to be delivered to a mammalian cell), it is typically referred to as "rAAV”.
  • heterologous polynucleotide i.e., a polynucleotide other than a wild-type AAV genome such as a transgene to be delivered to a mammalian cell
  • the AAV expression vectors are constructed using known techniques to at least provide as operatively linked components in the direction of transcription, control elements including a transcriptional initiation region, the DNA of interest and a transcriptional termination region.
  • the control elements are selected to be functional in a mammalian cell.
  • the resulting construct which contains the operatively linked components is flanked (5' and 3') with functional AAV ITR sequences.
  • AAV ITRs adeno-associated virus inverted terminal repeats
  • AAV ITRs the art-recognized regions found at each end of the AAV genome which function together in cis as origins of DNA replication and as packaging signals for the virus.
  • AAV ITRs, together with the AAV rep coding region, provide for the efficient excision and rescue from, and integration of a nucleotide sequence interposed between two flanking ITRs into a mammalian cell genome.
  • AAV ITR The nucleotide sequences of AAV ITR regions are known. As used herein, an "AAV ITR" need not have the wild-type nucleotide sequence depicted, but may be altered, e.g., by the insertion, deletion or substitution of nucleotides. Additionally, the AAV ITR may be derived from any of several AAV serotypes, including without limitation, AAV1, AAV2, AAV3, AAV4, AAV5, AAV7, etc.
  • 5' and 3' ITRs which flank a selected nucleotide sequence in an AAV vector need not necessarily be identical or derived from the same AAV serotype or isolate, so long as they function as intended, i.e., to allow for excision and rescue of the sequence of interest from a host cell genome or vector, and to allow integration of the heterologous sequence into the recipient cell genome when AAV Rep gene products are present in the cell.
  • AAV ITRs can be derived from any of several AAV serotypes, including without limitation, AAV1, AAV2, AAV3, AAV4, AAV5, AAV7, etc.
  • 5' and 3' ITRs which flank a selected nucleotide sequence in an AAV expression vector need not necessarily be identical or derived from the same AAV serotype or isolate, so long as they function as intended, i.e., to allow for excision and rescue of the sequence of interest from a host cell genome or vector, and to allow integration of the DNA molecule into the recipient cell genome when AAV Rep gene products are present in the cell.
  • AAV capsids can be derived from AAV2.
  • Suitable DNA molecules for use in AAV vectors will be less than about 5 kilobases (kb), less than about 4.5 kb, less than about 4kb, less than about 3.5 kb, less than about 3 kb, less than about 2.5 kb in size and are known in the art.
  • the selected nucleotide sequence is operably linked to control elements that direct the transcription or expression thereof in the subject in vivo.
  • control elements can comprise control sequences normally associated with the selected gene.
  • heterologous control sequences can be employed.
  • Useful heterologous control sequences generally include those derived from sequences encoding mammalian or viral genes.
  • Examples include, but are not limited to, the SV40 early promoter, mouse mammary tumor virus LTR promoter; adenovirus major late promoter (Ad MLP); a herpes simplex virus (HSV) promoter, a cytomegalovirus (CMV) promoter such as the CMV immediate early promoter region (CMVIE), a rous sarcoma virus (RSV) promoter, pol II promoters, pol III promoters, synthetic promoters, hybrid promoters, and the like.
  • CMVIE CMV immediate early promoter region
  • RSV rous sarcoma virus
  • sequences derived from nonviral genes such as the murine metallothionein gene, will also find use herein.
  • Such promoter sequences are commercially available from, e.g., Stratagene (San Diego, Calif.).
  • heterologous promoters and other control elements such as CNS-specific and inducible promoters, enhancers and the like
  • heterologous promoters include the CMV promoter.
  • CNS-specific promoters include those isolated from the genes from myelin basic protein (MBP), glial fibrillary acid protein (GFAP), and neuron specific enolase (NSE).
  • MBP myelin basic protein
  • GFAP glial fibrillary acid protein
  • NSE neuron specific enolase
  • inducible promoters include DNA responsive elements for ecdysone, tetracycline, hypoxia and aufin.
  • the AAV expression vector which harbors the DNA molecule of interest bounded by AAV ITRs can be constructed by directly inserting the selected sequence(s) into an AAV genome which has had the major AAV open reading frames ("ORFs") excised therefrom.
  • ORFs major AAV open reading frames
  • Other portions of the AAV genome can also be deleted, so long as a sufficient portion of the ITRs remain to allow for replication and packaging functions.
  • Such constructs can be designed using techniques well known in the art.
  • AAV ITRs can be excised from the viral genome or from an AAV vector containing the same and fused 5' and 3' of a selected nucleic acid construct that is present in another vector using standard ligation techniques.
  • ligations can be accomplished in 20 mM Tris-Cl pH 7.5, 10 mM MgCl 2 , 10 mM DTT, 33 ⁇ g/ml BSA, 10 mM-50 mM NaCl, and either 40 uM ATP, 0.01-0.02 (Weiss) units T4 DNA ligase at 0°C (for "sticky end” ligation) or 1 mM ATP, 0.3-0.6 (Weiss) units T4 DNA ligase at 14°C (for "blunt end” ligation). Intermolecular "sticky end” ligations are usually performed at 30-100 ⁇ g/ml total DNA concentrations (5-100 nM total end concentration).
  • chimeric genes can be produced synthetically to include AAV ITR sequences arranged 5' and 3' of one or more selected nucleic acid sequences. Preferred codons for expression of the chimeric gene sequence in mammalian CNS cells can be used. The complete chimeric sequence is assembled from overlapping oligonucleotides prepared by standard methods.
  • an AAV expression vector is introduced into a suitable host cell using known techniques, such as by transfection.
  • transfection techniques are generally known in the art. See, e.g., Sambrook et al. (1989) Molecular Cloning, a laboratory manual, Cold Spring Harbor Laboratories, New York .
  • Particularly suitable transfection methods include calcium phosphate co-precipitation, direct micro-injection into cultured cells, electroporation, liposome mediated gene transfer, lipid-mediated transduction, and nucleic acid delivery using high-velocity microprojectiles.
  • suitable host cells for producing rAAV virions include microorganisms, yeast cells, insect cells, and mammalian cells, that can be, or have been, used as recipients of a heterologous DNA molecule.
  • the term includes the progeny of the original cell which has been transfected.
  • a "host cell” as used herein generally refers to a cell which has been transfected with an exogenous DNA sequence. Cells from the stable human cell line, 293 (readily available through, e.g., the American Type Culture Collection under Accession Number ATCC CRL1573) can be used in the practice of the present disclosure.
  • the human cell line 293 is a human embryonic kidney cell line that has been transformed with adenovirus type-5 DNA fragments, and expresses the adenoviral E1a and E1b genes.
  • the 293 cell line is readily transfected, and provides a particularly convenient platform in which to produce rAAV virions.
  • AAV rep coding region is meant the art-recognized region of the AAV genome which encodes the replication proteins Rep 78, Rep 68, Rep 52 and Rep 40. These Rep expression products have been shown to possess many functions, including recognition, binding and nicking of the AAV origin of DNA replication, DNA helicase activity and modulation of transcription from AAV (or other heterologous) promoters. The Rep expression products are collectively required for replicating the AAV genome. Suitable homologues of the AAV rep coding region include the human herpesvirus 6 (HHV-6) rep gene which is also known to mediate AAV-2 DNA replication.
  • HHV-6 human herpesvirus 6
  • AAV cap coding region is meant the art-recognized region of the AAV genome which encodes the capsid proteins VP1, VP2, and VP3, or functional homologues thereof. These Cap expression products supply the packaging functions which are collectively required for packaging the viral genome.
  • AAV helper functions are introduced into the host cell by transfecting the host cell with an AAV helper construct either prior to, or concurrently with, the transfection of the AAV expression vector.
  • AAV helper constructs are thus used to provide at least transient expression of AAV rep and/or cap genes to complement missing AAV functions that are necessary for productive AAV infection.
  • AAV helper constructs lack AAV ITRs and can neither replicate nor package themselves. These constructs can be in the form of a plasmid, phage, transposon, cosmid, virus, or virion.
  • a number of AAV helper constructs have been described, such as the commonly used plasmids pAAV/Ad and pIM29+45 which encode both Rep and Cap expression products.
  • a number of other vectors have been described which encode Rep and/or Cap expression products.
  • Methods of delivery of viral vectors include injecting the AAV2 into the CSF.
  • rAAV virions may be introduced into cells of the CNS using either in vivo or in vitro transduction techniques. If transduced in vitro, the desired recipient cell will be removed from the subject, transduced with rAAV virions and reintroduced into the subject.
  • syngeneic or xenogeneic cells can be used where those cells will not generate an inappropriate immune response in the subject.
  • transduced cells can be transduced in vitro by combining recombinant AAV virions with CNS cells e.g., in appropriate media, and screening for those cells harboring the DNA of interest can be screened using conventional techniques such as Southern blots and/or PCR, or by using selectable markers.
  • Transduced cells can then be formulated into pharmaceutical compositions, described more fully below, and the composition introduced into the subject by various techniques, such as by grafting, intramuscular, intravenous, subcutaneous and intraperitoneal injection.
  • compositions will comprise sufficient genetic material to produce a therapeutically effective amount of the nucleic acid of interest, i.e., an amount sufficient to reduce or ameliorate symptoms of the disease state in question or an amount sufficient to confer the desired benefit.
  • the pharmaceutical compositions will also contain a pharmaceutically acceptable excipient.
  • excipients include any pharmaceutical agent that does not itself induce the production of antibodies harmful to the individual receiving the composition, and which may be administered without undue toxicity.
  • Pharmaceutically acceptable excipients include, but are not limited to, sorbitol, Tween80, and liquids such as water, saline, glycerol and ethanol.
  • Pharmaceutically acceptable salts can be included therein, for example, mineral acid salts such as hydrochlorides, hydrobromides, phosphates, sulfates, and the like; and the salts of organic acids such as acetates, propionates, malonates, benzoates, and the like. Additionally, auxiliary substances, such as wetting or emulsifying agents, pH buffering substances, and the like, may be present in such vehicles.
  • mineral acid salts such as hydrochlorides, hydrobromides, phosphates, sulfates, and the like
  • organic acids such as acetates, propionates, malonates, benzoates, and the like
  • auxiliary substances such as wetting or emulsifying agents, pH buffering substances, and the like, may be present in such vehicles.
  • an effective amount of viral vector which must be added can be empirically determined. Administration can be effected in one dose, continuously or intermittently throughout the course of treatment. Methods of determining the most effective means and dosages of administration are well known to those of skill in the art and will vary with the viral vector, the composition of the therapy, the target cells, and the subject being treated. Single and multiple administrations can be carried out with the dose level and pattern being selected by the treating physician.
  • transgene could be expressed by the delivered viral vector.
  • separate vectors, each expressing one or more different transgenes can also be delivered to the CNS as described herein.
  • viral vectors delivered by the methods of the present disclosure be combined with other suitable compositions and therapies.
  • the exogenous genetic material e.g ., a cDNA encoding one or more therapeutic proteins
  • the exogenous genetic material is introduced into the cell ex vivo or in vivo by genetic transfer methods, such as transfection or transduction, to provide a genetically modified cell.
  • Various expression vectors i.e., vehicles for facilitating delivery of exogenous genetic material into a target cell
  • transfection of cells refers to the acquisition by a cell of new genetic material by incorporation of added DNA.
  • transfection refers to the insertion of nucleic acid into a cell using physical or chemical methods.
  • transfection techniques are known to those of ordinary skill in the art including: calcium phosphate DNA co-precipitation; DEAE-dextran; electroporation; cationic liposome-mediated transfection; and tungsten particle-faciliated microparticle bombardment.
  • Strontium phosphate DNA co-precipitation is another possible transfection method.
  • transduction of cells refers to the process of transferring nucleic acid into a cell using a DNA or RNA virus.
  • a RNA virus i.e., a retrovirus
  • Exogenous genetic material contained within the retrovirus is incorporated into the genome of the transduced cell.
  • a cell that has been transduced with a chimeric DNA virus e.g. , an adenovirus carrying a cDNA encoding a therapeutic agent
  • the exogenous genetic material includes the heterologous gene (usually in the form of a cDNA comprising the exons coding for the therapeutic protein) together with a promoter to control transcription of the new gene.
  • the promoter characteristically has a specific nucleotide sequence necessary to initiate transcription.
  • the exogenous genetic material further includes additional sequences (i.e., enhancers) required to obtain the desired gene transcription activity.
  • enhancers i.e., enhancers
  • an “enhancer” is simply any non-translated DNA sequence which works contiguous with the coding sequence (in cis) to change the basal transcription level dictated by the promoter.
  • the exogenous genetic material may introduced into the cell genome immediately downstream from the promoter so that the promoter and coding sequence are operatively linked so as to permit transcription of the coding sequence.
  • a retroviral expression vector may include an exogenous promoter element to control transcription of the inserted exogenous gene.
  • exogenous promoters include both constitutive and inducible promoters.
  • constitutive promoters control the expression of essential cell functions. As a result, a gene under the control of a constitutive promoter is expressed under all conditions of cell growth.
  • Exemplary constitutive promoters include the promoters for the following genes which encode certain constitutive or "housekeeping" functions: hypoxanthine phosphoribosyl transferase (HPRT), dihydrofolate reductase (DHFR), adenosine deaminase, phosphoglycerol kinase (PGK), pyruvate kinase, phosphoglycerol mutase, the actin promoter, and other constitutive promoters known to those of skill in the art.
  • HPRT hypoxanthine phosphoribosyl transferase
  • DHFR dihydrofolate reductase
  • PGK phosphoglycerol kinase
  • pyruvate kinase phosphoglycerol mutase
  • viral promoters function constitutively in eucaryotic cells. These include: the early and late promoters of SV40; the long terminal repeats (LTRs) of Moloney Leukemia Virus and other retroviruses; and the thymidine kinase promoter of Herpes Simplex Virus, among many others. Accordingly, any of the above-referenced constitutive promoters can be used to control transcription of a heterologous gene insert.
  • inducible promoters Genes that are under the control of inducible promoters are expressed only or to a greater degree, in the presence of an inducing agent, (e.g. , transcription under control of the metallothionein promoter is greatly increased in presence of certain metal ions).
  • Inducible promoters include responsive elements (REs) which stimulate transcription when their inducing factors are bound.
  • REs responsive elements
  • Promoters containing a particular RE can be chosen in order to obtain an inducible response and in some cases, the RE itself may be attached to a different promoter, thereby conferring inducibility to the recombinant gene.
  • the appropriate promoter constitutive versus inducible; strong versus weak
  • delivery of the therapeutic agent in situ is triggered by exposing the genetically modified cell in situ to conditions for permitting transcription of the therapeutic agent, e.g., by intraperitoneal injection of specific inducers of the inducible promoters which control transcription of the agent.
  • in situ expression by genetically modified cells of a therapeutic agent encoded by a gene under the control of the metallothionein promoter is enhanced by contacting the genetically modified cells with a solution containing the appropriate (i.e., inducing) metal ions in situ.
  • the amount of therapeutic agent that is delivered in situ is regulated by controlling such factors as: (1) the nature of the promoter used to direct transcription of the inserted gene, (i.e., whether the promoter is constitutive or inducible, strong or weak); (2) the number of copies of the exogenous gene that are inserted into the cell; (3) the number of transduced/transfected cells that are administered ( e.g. , implanted) to the patient; (4) the size of the implant ( e.g. , graft or encapsulated expression system); (5) the number of implants; (6) the length of time the transduced/transfected cells or implants are left in place; and (7) the production rate of the therapeutic agent by the genetically modified cell. Selection and optimization of these factors for delivery of a therapeutically effective dose of a particular therapeutic agent is deemed to be within the scope of one of ordinary skill in the art without undue experimentation, taking into account the above-disclosed factors and the clinical profile of the patient.
  • the expression vector may include a selection gene, for example, a neomycin resistance gene, for facilitating selection of cells that have been transfected or transduced with the expression vector.
  • the cells are transfected with two or more expression vectors, at least one vector containing the gene(s) encoding the therapeutic agent(s), the other vector containing a selection gene.
  • a suitable promoter, enhancer, selection gene and/or signal sequence (described below) is deemed to be within the scope of one of ordinary skill in the art without undue experimentation.
  • the therapeutic agent can be targeted for delivery to an extracellular, intracellular or membrane location.
  • the expression vector is designed to include an appropriate secretion "signal" sequence for secreting the therapeutic gene product from the cell to the extracellular milieu. If it is desirable for the gene product to be retained within the cell, this secretion signal sequence is omitted.
  • the expression vector can be constructed to include "retention" signal sequences for anchoring the therapeutic agent within the cell plasma membrane. For example, all membrane proteins have hydrophobic transmembrane regions, which stop translocation of the protein in the membrane and do not allow the protein to be secreted. The construction of an expression vector including signal sequences for targeting a gene product to a particular location is deemed to be within the scope of one of ordinary skill in the art without the need for undue experimentation.
  • LSDs Lysosomal storage disorders
  • CNS central nervous system
  • LINCL Late infantile neuronal ceroid lipofuscinosis
  • TPP1 lysosomal protease tripeptidyl peptidase 1
  • ERT Enzyme-replacement therapy
  • the next step was to determine whether long-term, steady-state levels of therapeutic enzymes could be achieved in a mammal. It was discovered that ependymal cells (cells that lie the ventricles in the brain) can be transduced and secrete a targeted enzyme into the cerebral spinal fluid (CSF). It was determined that adeno-associated virus (AAV4) can transduce the ependyma in a mouse model with high efficiency. Davidson et al, PNAS, 28:3428-3432, 2000 . It was found that in mice there was a normalization of stored substrate levels in disease brain after AAV4 treatment.
  • AAV4 adeno-associated virus
  • a vector needed to be found that could transduce ependymal cells (cells that line the ventricles) in the brain of larger mammals. Studies were performed in a dog model of LINCL and a non-human primate model of LINCL. The LINCL dogs are normal at birth, but develop neurological signs around 7 months, testable cognitive deficits at ⁇ 5-6 months, seizures at 10-11 months, and progressive visual loss.
  • AAV adeno-associated virus
  • AAV2/1 i.e., AAV2 ITR and AAV1 capsid
  • AAV2/2, AAV2/4, AAV2/5, and AAV2/8 AAV2/2 worked much better in the large mammals (dogs and NHP), followed by AAV2/8, AAV2/5, AAV2/1 and AAV2/4. This was quite surprising because the order of effectiveness of the viral vectors is the opposite of what was observed in mice.
  • ventricular lining cells can be a source of recombinant enzyme in CSF for distribution throughout the brain, and that AAV2/2 is an effective vehicle for administering therapeutic agents, such as the gene encoding CLN2 (TPP1) in dogs and nonhuman primates.
  • therapeutic agents such as the gene encoding CLN2 (TPP1) in dogs and nonhuman primates.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Zoology (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Wood Science & Technology (AREA)
  • Molecular Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • General Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • Epidemiology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Physics & Mathematics (AREA)
  • Biophysics (AREA)
  • Plant Pathology (AREA)
  • Virology (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Claims (4)

  1. rAAV2-Partikel, umfassend ein AAV2-Kapsidprotein und einen Vektor, der eine Nukleinsäure, die für ein therapeutisches Protein kodiert, die zwischen einem Paar von invertierten terminalen Wiederholungen von AAV eingefügt ist, umfasst, zur Verwendung bei der Behandlung einer lysosomalen Speicherkrankheit (LSK) in einem Säugetier, das kein Nagetier ist, wobei der rAAV2-Partikel dem Säugetier, das kein Nagetier ist, durch intraventrikuläre Injektion oder ICV-Gabe verabreicht werden soll, wobei das Protein Tripeptidylpeptidase 1 (TPP1) ist, wobei die LSK spätinfantile neuronale Ceroid-Lipofuszinose (LINCL) ist, und wobei das Säugetier, das kein Nagetier ist, ein Hund, Primat oder Mensch ist.
  2. rAAV2-Partikel, umfassend ein AAV2-Kapsidprotein und einen Vektor, der eine Nukleinsäure, die für ein therapeutisches Protein kodiert, die zwischen einem Paar von invertierten terminalen Wiederholungen von AAV eingefügt ist, umfasst, zur Verwendung gemäß Anspruch 1, wobei das Säugetier, das kein Nagetier ist, ein Primat oder Hund ist.
  3. rAAV2-Partikel, umfassend ein AAV2-Kapsidprotein und einen Vektor, der eine Nukleinsäure, die für ein therapeutisches Protein kodiert, die zwischen einem Paar von invertierten terminalen Wiederholungen von AAV eingefügt ist, umfasst, zur Verwendung gemäß Anspruch 1, wobei das Säugetier, das kein Nagetier ist, ein Mensch ist.
  4. rAAV2-Partikel, umfassend ein AAV2-Kapsidprotein und einen Vektor, der eine Nukleinsäure, die für ein therapeutisches Protein kodiert, die zwischen einem Paar von invertierten terminalen Wiederholungen von AAV eingefügt ist, umfasst, zur Verwendung gemäß Anspruch 1, wobei das Paar von invertierten terminalen Wiederholungen von AAV ein Paar von invertierten terminalen Wiederholungen von AAV2 ist.
EP12763234.7A 2011-03-31 2012-04-02 AAV2-Partikel, umfassend ein AAV2-Capsidprotein und einen Vektor, umfassend eine Nukleinsäure, die eine Tripeptidylpeptidase 1 (TPP1) kodiert, zur Verwendung für die Behandlung von spätinfaniler Ceroid-Lipofuszinose (LINCL) in einem Nicht-Nagetier-Säugetier durch intraventrikuläre Injektion oder ICV-Gabe. Active EP2691529B1 (de)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP19170680.3A EP3546584B1 (de) 2011-03-31 2012-04-02 Zusammensetzungen zur behandlung lysosomaler speichererkrankung

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201161470460P 2011-03-31 2011-03-31
PCT/US2012/031896 WO2012135857A1 (en) 2011-03-31 2012-04-02 Methods and compositions for treating brain diseases

Related Child Applications (2)

Application Number Title Priority Date Filing Date
EP19170680.3A Division EP3546584B1 (de) 2011-03-31 2012-04-02 Zusammensetzungen zur behandlung lysosomaler speichererkrankung
EP19170680.3A Division-Into EP3546584B1 (de) 2011-03-31 2012-04-02 Zusammensetzungen zur behandlung lysosomaler speichererkrankung

Publications (3)

Publication Number Publication Date
EP2691529A1 EP2691529A1 (de) 2014-02-05
EP2691529A4 EP2691529A4 (de) 2014-12-10
EP2691529B1 true EP2691529B1 (de) 2019-06-12

Family

ID=46931978

Family Applications (2)

Application Number Title Priority Date Filing Date
EP19170680.3A Active EP3546584B1 (de) 2011-03-31 2012-04-02 Zusammensetzungen zur behandlung lysosomaler speichererkrankung
EP12763234.7A Active EP2691529B1 (de) 2011-03-31 2012-04-02 AAV2-Partikel, umfassend ein AAV2-Capsidprotein und einen Vektor, umfassend eine Nukleinsäure, die eine Tripeptidylpeptidase 1 (TPP1) kodiert, zur Verwendung für die Behandlung von spätinfaniler Ceroid-Lipofuszinose (LINCL) in einem Nicht-Nagetier-Säugetier durch intraventrikuläre Injektion oder ICV-Gabe.

Family Applications Before (1)

Application Number Title Priority Date Filing Date
EP19170680.3A Active EP3546584B1 (de) 2011-03-31 2012-04-02 Zusammensetzungen zur behandlung lysosomaler speichererkrankung

Country Status (7)

Country Link
US (1) US9849195B2 (de)
EP (2) EP3546584B1 (de)
CA (1) CA2832151C (de)
DK (1) DK2691529T3 (de)
ES (1) ES2745470T3 (de)
PT (1) PT2691529T (de)
WO (1) WO2012135857A1 (de)

Families Citing this family (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014186579A1 (en) 2013-05-15 2014-11-20 Regents Of The University Of Minnesota Adeno-associated virus mediated gene transfer to the central nervous system
CN105764532B (zh) * 2013-07-26 2021-08-31 衣阿华大学研究基金会 治疗脑疾病的方法和组合物
EP3071240B1 (de) * 2013-11-20 2021-01-06 University of Iowa Research Foundation Verfahren zur überwachung der wirksamkeit der behandlung von alzheimer mit einer apoe isoform
GB201508025D0 (en) 2015-05-11 2015-06-24 Ucl Business Plc Fabry disease gene therapy
ES2901766T3 (es) * 2015-05-15 2022-03-23 Regenxbio Inc Virus adenoasociado para el suministro terapéutico al sistema nervioso central
JP7190352B2 (ja) * 2015-10-23 2022-12-15 ユニバーシティー オブ アイオワ リサーチ ファウンデーション 認知保護を提供しつつ疾患の発症及び進行を遅らせるための遺伝子治療を用いた神経変性疾患の治療方法
US10927155B2 (en) 2015-11-19 2021-02-23 George Mason Research Foundation, Inc. Dual inhibitory action peptidomimetic inhibitor for IL-33 and IL-1beta
WO2017181113A1 (en) 2016-04-15 2017-10-19 The Trustees Of The University Of Pennsyvania Gene therapy for treating mucopolysaccharidosis type ii
BR112019009074A2 (pt) * 2016-11-04 2019-07-16 Childrens Hospital Philadelphia composições, métodos e uso de transferência genética para o tratamento de doenças neurodegenerativas
RU2019140862A (ru) * 2017-05-12 2021-06-15 Дзе Чилдрен'З Хоспитал Оф Филадельфия Варианты сульфамидазы (sgsh), векторы, композиции и способы, и применения для лечения мукополисахаридоз типа iiia (mps iiia)
CN110809583A (zh) 2017-06-07 2020-02-18 瑞泽恩制药公司 用于内化酶的组合物和方法
SG11202002457RA (en) 2017-09-22 2020-04-29 Univ Pennsylvania Gene therapy for treating mucopolysaccharidosis type ii
SG11202104295UA (en) * 2018-11-14 2021-06-29 Regenxbio Inc Gene therapy for neuronal ceroid lipofuscinoses

Family Cites Families (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ATE384787T1 (de) 1994-11-08 2008-02-15 Novozymes As Tripeptidyl-aminopeptidase
US20020037867A1 (en) 1999-02-26 2002-03-28 James M. Wilson Method for recombinant adeno-associated virus-directed gene therapy
US6302685B1 (en) 1997-09-16 2001-10-16 University Of Medicine And Dentistry Of New Jersey Human lysosomal protein and methods of its use
US6855314B1 (en) 2000-03-22 2005-02-15 The United States Of America As Represented By The Department Of Health And Human Services AAV5 vector for transducing brain cells and lung cells
US6468524B1 (en) 2000-03-22 2002-10-22 The United States Of America, As Represented By The Secretary Of The Department Of Health And Human Services AAV4 vector and uses thereof
US7749971B2 (en) 2001-02-20 2010-07-06 Bedwell David M Aminoglycoside treatment for lysosomal storage diseases
CA2542417A1 (en) * 2003-10-14 2005-04-28 Neurologix Research, Inc. Methods and compositions for the treatment of neurological disease
WO2008049058A2 (en) 2006-10-18 2008-04-24 Cornell Research Foundation, Inc. Cln2 treatment of alzheimer's disease
SI2154969T1 (sl) 2007-05-16 2016-02-29 The Brigham And Women's Hospital, Inc. Zdravljenje sinukleinopatij
ES2905616T3 (es) * 2007-06-06 2022-04-11 Genzyme Corp Terapia génica para enfermedades de almacenamiento lisosómico
ES2615180T3 (es) * 2007-07-14 2017-06-05 University Of Iowa Research Foundation Métodos y composiciones para el tratamiento de enfermedades cerebrales
WO2014089449A1 (en) 2012-12-07 2014-06-12 Rush University Nedical Center Composition and method for treating neuronal ceroid lipofuscinosis
GB201308917D0 (en) 2013-05-17 2013-07-03 Renishaw Plc Delivery

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
COOK AARON M ET AL: "Intracerebroventricular administration of drugs", PHARMACOTHERAPY : THE JOURNAL OF HUMAN PHARMACOLOGY AND DRUG THERAPY, WILEY-BLACKWELL, US, vol. 29, no. 7, 1 July 2009 (2009-07-01), pages 832 - 845, XP009147777, ISSN: 0277-0008, DOI: 10.1592/PHCO.29.7.832 *
PASSINI M A ET AL: "Intraventricular brain injection of adeno-associated virus type 1(AAV1) in neonatal mice results in complementary patterns of neuronal transduction to AAV2 and total long-term correction of storage lesions in the brains of beta-glucuronidase-deficient mice", JOURNAL OF VIROLOGY, THE AMERICAN SOCIETY FOR MICROBIOLOGY, US, vol. 77, no. 12, 1 June 2003 (2003-06-01), pages 7034 - 7040, XP002953219, ISSN: 0022-538X, DOI: 10.1128/JVI.77.12.7034-7040.2003 *
PASSINI MARCO A ET AL: "Intracranial delivery of CLN2 reduces brain pathology in a mouse model of classical late infantile neuronal ceroid lipofuscinosis", JOURNAL OF NEUROSCI, THE SOCIETY, WASHINGTON, DC, US, vol. 26, no. 5, 1 February 2006 (2006-02-01), pages 1334 - 1342, XP009184135, ISSN: 1529-2401, DOI: 10.1523/JNEUROSCI.2676-05.2006 *

Also Published As

Publication number Publication date
CA2832151A1 (en) 2012-10-04
WO2012135857A9 (en) 2012-10-26
US9849195B2 (en) 2017-12-26
US20140088179A1 (en) 2014-03-27
ES2745470T3 (es) 2020-03-02
EP2691529A1 (de) 2014-02-05
DK2691529T3 (da) 2019-09-23
EP3546584A1 (de) 2019-10-02
PT2691529T (pt) 2019-09-27
EP3546584B1 (de) 2024-02-14
WO2012135857A1 (en) 2012-10-04
EP2691529A4 (de) 2014-12-10
CA2832151C (en) 2021-06-15

Similar Documents

Publication Publication Date Title
US20200009267A1 (en) Methods and compositions for treating brain diseases
EP2691529B1 (de) AAV2-Partikel, umfassend ein AAV2-Capsidprotein und einen Vektor, umfassend eine Nukleinsäure, die eine Tripeptidylpeptidase 1 (TPP1) kodiert, zur Verwendung für die Behandlung von spätinfaniler Ceroid-Lipofuszinose (LINCL) in einem Nicht-Nagetier-Säugetier durch intraventrikuläre Injektion oder ICV-Gabe.
JP6873907B2 (ja) 遺伝性眼疾患を治療するための方法及び組成物
US8691948B2 (en) Methods and compositions for treating brain diseases
CN116670159A (zh) 组合物及其用于治疗安格尔曼综合征的用途
EP4259790A1 (de) Varianten lysosomaler saurer lipase und verwendungen davon

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20131029

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

DAX Request for extension of the european patent (deleted)
A4 Supplementary search report drawn up and despatched

Effective date: 20141112

RIC1 Information provided on ipc code assigned before grant

Ipc: C12N 9/48 20060101ALI20141106BHEP

Ipc: A61K 48/00 20060101ALI20141106BHEP

Ipc: C12N 15/86 20060101AFI20141106BHEP

17Q First examination report despatched

Effective date: 20160714

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

INTG Intention to grant announced

Effective date: 20180713

GRAJ Information related to disapproval of communication of intention to grant by the applicant or resumption of examination proceedings by the epo deleted

Free format text: ORIGINAL CODE: EPIDOSDIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

INTC Intention to grant announced (deleted)
RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: UNIVERSITY OF IOWA RESEARCH FOUNDATION

RIN1 Information on inventor provided before grant (corrected)

Inventor name: DAVIDSON, BEVERLY, L.

INTG Intention to grant announced

Effective date: 20181205

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE PATENT HAS BEEN GRANTED

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 1142592

Country of ref document: AT

Kind code of ref document: T

Effective date: 20190615

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602012060940

Country of ref document: DE

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: DK

Ref legal event code: T3

Effective date: 20190916

REG Reference to a national code

Ref country code: NL

Ref legal event code: FP

REG Reference to a national code

Ref country code: PT

Ref legal event code: SC4A

Ref document number: 2691529

Country of ref document: PT

Date of ref document: 20190927

Kind code of ref document: T

Free format text: AVAILABILITY OF NATIONAL TRANSLATION

Effective date: 20190910

REG Reference to a national code

Ref country code: SE

Ref legal event code: TRGR

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG4D

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: HR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190612

Ref country code: AL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190612

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190612

REG Reference to a national code

Ref country code: NO

Ref legal event code: T2

Effective date: 20190612

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190912

Ref country code: RS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190612

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190913

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190612

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190612

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190612

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190612

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SM

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190612

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20191012

REG Reference to a national code

Ref country code: ES

Ref legal event code: FG2A

Ref document number: 2745470

Country of ref document: ES

Kind code of ref document: T3

Effective date: 20200302

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602012060940

Country of ref document: DE

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: TR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190612

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190612

26N No opposition filed

Effective date: 20200313

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200224

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190612

PG2D Information on lapse in contracting state deleted

Ref country code: IS

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190612

REG Reference to a national code

Ref country code: AT

Ref legal event code: UEP

Ref document number: 1142592

Country of ref document: AT

Kind code of ref document: T

Effective date: 20190612

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190612

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190612

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190612

P01 Opt-out of the competence of the unified patent court (upc) registered

Effective date: 20230601

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: NO

Payment date: 20230427

Year of fee payment: 12

Ref country code: IT

Payment date: 20230419

Year of fee payment: 12

Ref country code: IE

Payment date: 20230427

Year of fee payment: 12

Ref country code: FR

Payment date: 20230425

Year of fee payment: 12

Ref country code: ES

Payment date: 20230503

Year of fee payment: 12

Ref country code: DK

Payment date: 20230427

Year of fee payment: 12

Ref country code: DE

Payment date: 20230427

Year of fee payment: 12

Ref country code: CH

Payment date: 20230502

Year of fee payment: 12

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: SE

Payment date: 20230427

Year of fee payment: 12

Ref country code: FI

Payment date: 20230425

Year of fee payment: 12

Ref country code: AT

Payment date: 20230321

Year of fee payment: 12

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: BE

Payment date: 20230427

Year of fee payment: 12

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GB

Payment date: 20230427

Year of fee payment: 12

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: CZ

Payment date: 20240321

Year of fee payment: 13

Ref country code: PT

Payment date: 20240322

Year of fee payment: 13

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: LU

Payment date: 20240429

Year of fee payment: 13

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: NL

Payment date: 20240426

Year of fee payment: 13